Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Diversification of TAM receptor tyrosine kinase function

Abstract

The clearance of apoptotic cells is critical for both tissue homeostasis and the resolution of inflammation. We found that the TAM receptor tyrosine kinases Axl and Mer had distinct roles as phagocytic receptors in these two settings, in which they exhibited divergent expression, regulation and activity. Mer acted as a tolerogenic receptor in resting macrophages and during immunosuppression. In contrast, Axl was an inflammatory response receptor whose expression was induced by proinflammatory stimuli. Axl and Mer differed in their ligand specificities, ligand-receptor complex formation in tissues, and receptor shedding upon activation. These differences notwithstanding, phagocytosis by either protein was strictly dependent on receptor activation triggered by bridging of TAM receptor–ligand complexes to the 'eat-me' signal phosphatidylserine on the surface of apoptotic cells.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Differences in the expression and activation of Axl and Mer.
Figure 2: The expression of Axl and Mer in inflammatory macrophages.
Figure 3: Axl is a phagocytic receptor in activated macrophages.
Figure 4: Axl and Mer kinase activity is necessary for the phagocytosis of apoptotic cells.
Figure 5: GAS-6 is bound to Axl in vivo and in vitro.
Figure 6: Axl- and Mer-activating antibodies.

Similar content being viewed by others

References

  1. Muñoz, L.E., Lauber, K., Schiller, M., Manfredi, A.A. & Herrmann, M. The role of defective clearance of apoptotic cells in systemic autoimmunity. Nat. Rev. Rheumatol. 6, 280–289 (2010).

    PubMed  Google Scholar 

  2. Lemke, G. Biology of the TAM receptors. Cold Spring Harbor Perspectives 5, a009076 (2013).

    Google Scholar 

  3. Lemke, G. & Rothlin, C.V. Immunobiology of the TAM receptors. Nat. Rev. Immunol. 8, 327–336 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Lu, Q. & Lemke, G. Homeostatic regulation of the immune system by receptor tyrosine kinases of the Tyro 3 family. Science 293, 306–311 (2001).

    CAS  PubMed  Google Scholar 

  5. Rothlin, C.V., Ghosh, S., Zuniga, E.I., Oldstone, M.B. & Lemke, G. TAM receptors are pleiotropic inhibitors of the innate immune response. Cell 131, 1124–1136 (2007).

    CAS  PubMed  Google Scholar 

  6. Burstyn-Cohen, T. et al. Genetic dissection of TAM receptor-ligand interaction in retinal pigment epithelial cell phagocytosis. Neuron 76, 1123–1132 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Scott, R.S. et al. Phagocytosis and clearance of apoptotic cells is mediated by MER. Nature 411, 207–211 (2001).

    CAS  PubMed  Google Scholar 

  8. Lemke, G. & Burstyn-Cohen, T. TAM receptors and the clearance of apoptotic cells. Ann. NY Acad. Sci. 1209, 23–29 (2010).

    CAS  PubMed  Google Scholar 

  9. Lu, Q. et al. Tyro-3 family receptors are essential regulators of mammalian spermatogenesis. Nature 398, 723–728 (1999).

    CAS  PubMed  Google Scholar 

  10. Bhattacharyya, S. et al. Enveloped viruses disable innate immune responses in dendritic cells by direct activation of TAM receptors. Cell Host Microbe 14, 136–147 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Meertens, L. et al. TIM and TAM receptors mediate dengue virus infection. Cell Host Microbe 12, 544–557 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Paolino, M. et al. Essential role of E3 ubiquitin ligase activity in Cbl-b-regulated T cell functions. J. Immunol. 186, 2138–2147 (2011).

    CAS  PubMed  Google Scholar 

  13. Schlegel, J. et al. MERTK receptor tyrosine kinase is a therapeutic target in melanoma. J. Clin. Invest. 123, 2257–2267 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Meyer, A.S., Miller, M.A., Gertler, F.B. & Lauffenburger, D.A. The receptor AXL diversifies EGFR signaling and limits the response to EGFR-targeted inhibitors in triple-negative breast cancer cells. Sci. Signal. 6, ra66 (2013).

    PubMed  PubMed Central  Google Scholar 

  15. Carrera Silva, E.A. et al. T cell-derived protein S engages TAM receptor signaling in dendritic cells to control the magnitude of the immune response. Immunity 39, 160–170 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Inaba, K. et al. Isolation of dendritic cells. Curr. Protoc. Immunol. 25, 3.7 (2009).

    Google Scholar 

  17. Rahman, Z.S., Shao, W.H., Khan, T.N., Zhen, Y. & Cohen, P.L. Impaired apoptotic cell clearance in the germinal center by Mer-deficient tingible body macrophages leads to enhanced antibody-forming cell and germinal center responses. J. Immunol. 185, 5859–5868 (2010).

    CAS  PubMed  Google Scholar 

  18. Subramanian, M. et al. An AXL/LRP-1/RANBP9 complex mediates DC efferocytosis and antigen cross-presentation in vivo. J. Clin. Invest. 124, 1296–308 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. McColl, A. et al. Glucocorticoids induce protein S-dependent phagocytosis of apoptotic neutrophils by human macrophages. J. Immunol. 183, 2167–2175 (2009).

    CAS  PubMed  Google Scholar 

  20. A-Gonzalez, N. et al. Apoptotic cells promote their own clearance and immune tolerance through activation of the nuclear receptor LXR. Immunity 31, 245–258 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Mukundan, L. et al. PPAR-delta senses and orchestrates clearance of apoptotic cells to promote tolerance. Nat. Med. 15, 1266–1272 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Clark, A.R. Anti-inflammatory functions of glucocorticoid-induced genes. Mol. Cell. Endocrinol. 275, 79–97 (2007).

    CAS  PubMed  Google Scholar 

  23. Mosser, D.M. & Edwards, J.P. Exploring the full spectrum of macrophage activation. Nat. Rev. Immunol. 8, 958–969 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Feng, X. et al. Lipopolysaccharide inhibits macrophage phagocytosis of apoptotic neutrophils by regulating the production of tumour necrosis factor alpha and growth arrest-specific gene 6. Immunology 132, 287–295 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Nishi, C., Toda, S., Segawa, K. & Nagata, S. Tim4- and MerTK- mediated engulfment of apoptotic cells by mouse resident peritoneal macrophages. Mol. Cell. Biol. 34, 1512–1520 (2014).

    PubMed  PubMed Central  Google Scholar 

  26. Seitz, H.M., Camenisch, T.D., Lemke, G., Earp, H.S. & Matsushima, G.K. Macrophages and dendritic cells use different Axl/Mertk/Tyro3 receptors in clearance of apoptotic cells. J. Immunol. 178, 5635–5642 (2007).

    CAS  PubMed  Google Scholar 

  27. Miksa, M., Komura, H., Wu, R., Shah, K.G. & Wang, P. A novel method to determine the engulfment of apoptotic cells by macrophages using pHrodo succinimidyl ester. J. Immunol. Methods 342, 71–77 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Oka, K. et al. Lectin-like oxidized low-density lipoprotein receptor 1 mediates phagocytosis of aged/apoptotic cells in endothelial cells. Proc. Natl. Acad. Sci. USA 95, 9535–9540 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Park, D. et al. BAI1 is an engulfment receptor for apoptotic cells upstream of the ELMO/Dock180/Rac module. Nature 450, 430–434 (2007).

    CAS  PubMed  Google Scholar 

  30. Schroeder, G.M. et al. Discovery of N-(4-(2-amino-3-chloropyridin-4-yloxy)-3-fluorophenyl)-4-ethoxy-1-(4-fluorophenyl)-2-oxo-1,2-dihydropyridine-3-carboxamide (BMS-777607), a selective and orally efficacious inhibitor of the Met kinase superfamily. J. Med. Chem. 52, 1251–1254 (2009).

    CAS  PubMed  Google Scholar 

  31. Lemmon, M.A. & Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 141, 1117–1134 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Kahn, C.R., Baird, K.L., Jarrett, D.B. & Flier, J.S. Direct demonstration that receptor crosslinking or aggregation is important in insulin action. Proc. Natl. Acad. Sci. USA 75, 4209–4213 (1978).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Schreiber, A.B., Libermann, T.A., Lax, I., Yarden, Y. & Schlessinger, J. Biological role of epidermal growth factor-receptor clustering. Investigation with monoclonal anti-receptor antibodies. J. Biol. Chem. 258, 846–853 (1983).

    CAS  PubMed  Google Scholar 

  34. Todt, J.C., Hu, B. & Curtis, J.L. The receptor tyrosine kinase MerTK activates phospholipase C gamma2 during recognition of apoptotic thymocytes by murine macrophages. J. Leukoc. Biol. 75, 705–713 (2004).

    CAS  PubMed  Google Scholar 

  35. O'Bryan, J.P., Fridell, Y.W., Koski, R., Varnum, B. & Liu, E.T. The transforming receptor tyrosine kinase, Axl, is post-translationally regulated by proteolytic cleavage. J. Biol. Chem. 270, 551–557 (1995).

    CAS  PubMed  Google Scholar 

  36. Costa, M., Bellosta, P. & Basilico, C. Cleavage and release of a soluble form of the receptor tyrosine kinase ARK in vitro and in vivo. J. Cell. Physiol. 168, 737–744 (1996).

    CAS  PubMed  Google Scholar 

  37. Ekman, C., Site, D.F., Gottsater, A., Lindblad, B. & Dahlback, B. Plasma concentrations of growth arrest specific protein 6 and the soluble form of its tyrosine kinase receptor Axl as markers of large abdominal aortic aneurysms. Clin. Biochem. 43, 110–114 (2010).

    CAS  PubMed  Google Scholar 

  38. Zhu, H. et al. Different expression patterns and clinical significance of mAxl and sAxl in systemic lupus erythematosus. Lupus 23, 624–634 (2014).

    CAS  PubMed  Google Scholar 

  39. Ko, C.P., Yu, Y.L., Hsiao, P.C., Yang, S.F. & Yeh, C.B. Plasma levels of soluble Axl correlate with severity of community-acquired pneumonia. Mol. Med. Rep. 9, 1400–1404 (2014).

    CAS  PubMed  Google Scholar 

  40. Liu, X. et al. Plasma concentrations of sAxl are associated with severe preeclampsia. Clin. Biochem. 47, 173–176 (2014).

    CAS  PubMed  Google Scholar 

  41. Lee, C.H. et al. Plasma concentrations predict aortic expression of growth-arrest-specific protein 6 in patients undergoing coronary artery bypass grafting. PLoS ONE 8, e79452 (2013).

    PubMed  PubMed Central  Google Scholar 

  42. Hsiao, F.C. et al. Circulating growth arrest-specific 6 protein is associated with adiposity, systemic inflammation, and insulin resistance among overweight and obese adolescents. J. Clin. Endocrinol. Metab. 98, E267–E274 (2013).

    CAS  PubMed  Google Scholar 

  43. Camenisch, T.D., Koller, B.H., Earp, H.S. & Matsushima, G.K. A novel receptor tyrosine kinase, Mer, inhibits TNF-alpha production and lipopolysaccharide-induced endotoxic shock. J. Immunol. 162, 3498–3503 (1999).

    CAS  PubMed  Google Scholar 

  44. Sen, P. et al. Apoptotic cells induce Mer tyrosine kinase-dependent blockade of NF-κB activation in dendritic cells. Blood 109, 653–660 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Casanova-Acebes, M. et al. Rhythmic modulation of the hematopoietic niche through neutrophil clearance. Cell 153, 1025–1035 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Scheiermann, C., Kunisaki, Y. & Frenette, P.S. Circadian control of the immune system. Nat. Rev. Immunol. 13, 190–198 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Holland, S.J. et al. R428, a selective small molecule inhibitor of Axl kinase, blocks tumor spread and prolongs survival in models of metastatic breast cancer. Cancer Res. 70, 1544–1554 (2010).

    CAS  PubMed  Google Scholar 

  48. Ye, X. et al. An anti-Axl monoclonal antibody attenuates xenograft tumor growth and enhances the effect of multiple anticancer therapies. Oncogene 29, 5254–5264 (2010).

    CAS  PubMed  Google Scholar 

  49. Rothlin, C.V. & Lemke, G. TAM receptor signaling and autoimmune disease. Curr. Opin. Immunol. 22, 740–746 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. van den Brand, B.T. et al. Therapeutic efficacy of Tyro3, Axl, and Mer tyrosine kinase agonists in collagen-induced arthritis. Arthritis Rheum. 65, 671–680 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Angelillo-Scherrer, A. et al. Deficiency or inhibition of Gas6 causes platelet dysfunction and protects mice against thrombosis. Nat. Med. 7, 215–221 (2001).

    CAS  PubMed  Google Scholar 

  52. Zhang, X., Goncalves, R. & Mosser, D.M. The isolation and characterization of murine macrophages. Curr. Protoc. Immunol. 83, 14.1 (2008).

    Google Scholar 

  53. Fourgeaud, L. et al. The metabotropic glutamate receptor mGluR5 is endocytosed by a clathrin-independent pathway. J. Biol. Chem. 278, 12222–12230 (2003).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Joens and J. Fitzpatrick for processing samples and acquiring scanning electron microscopic images; R. Evans (Salk Institute) for nuclear receptor agonists T0901317, GW501516 and BRL49653; J. Hash, P. Burrola and C. Mayer for technical support; and members of the Lemke laboratory and the Nomis Center for discussions. Supported by the US National Institutes of Health (R01 AI077058, R01 AI101400 and R01 NS085296 to G.L.), the Leona M. and Harry B. Helmsley Charitable Trust (2012-PG-MED002 to G.L.) the Nomis Foundation, the H.N. and Frances C. Berger Foundation, the Fritz B. Burns Foundation, the HKT Foundation, Françoise Gilot-Salk, the Human Frontiers Science Program (A.Z.), the Marie Curie Seventh Framework Programme (P.G.T.), the Leukemia and Lymphoma Society and the Nomis Foundation (E.D.L.).

Author information

Authors and Affiliations

Authors

Contributions

A.Z. designed and performed the experiments; P.G.T. aided in the design and execution of in vivo experiments; E.D.L. prepared purified recombinant GAS-6; I.D. aided in the design of the flow-cytometry-based phagocytosis assay; G.L. contributed to the design of the experiments and wrote the manuscript.

Corresponding author

Correspondence to Greg Lemke.

Ethics declarations

Competing interests

G.L. is a shareholder in Kolltan Pharmaceuticals.

Integrated supplementary information

Supplementary Figure 1 Expression of Axl and Mer on populations of cells of the immune system in vivo.

Immunohistochemistry of Axl and Mer in spleen (a), liver (b), and lung (c). Closed arrowheads: cells co-expressing Axl and Mer; open arrowheads: cells expressing mostly Axl. In the spleen, the principal Axl-Mer co-expressing cells are F4/80+ red pulp (RP) macrophages. In the splenic white pulp (WP) (d), Tingible body macrophages (F4/80-CD68+) express Mer and low levels of Axl (closed arrowheads), whereas a subpopulation of splenic CD11c+ DCs express only Axl (open arrowheads). In the lung (c and e), alveolar macrophages (CD11c+CD11b-MHCII-F4/80lo) are only Axl-positive (open arrowheads). In the liver, F4/80+ Kupffer cells are both Axl and Mer positive (b, closed arrowheads). Bars, 50 μm (a-c); 10 μm (d); and 20 μm (e). Representative images from n=3 mice.

Supplementary Figure 2 Discrete Axl+ and Mer+ cell populations in vitro.

(a) Unstimulated (Ctrl) or IFN-γ (250 U/ml, 18 h) treated BMDM cultures were stained live with Mer (green) and Axl (magenta) antibodies. Closed arrows: cells expressing mostly Axl; open arrows- cells expressing mostly Mer. The asterisk marks a single cell that is weakly positive for both Axl and Mer. Bar, 20 μm. Representative images of three independent experiments.

(b,c) BMDM cultures were stimulated with 0.1 μM Dex or 10 μg/ml poly(I:C) for 24 h, fixed and stained with Mer (b) or Axl (c) antibodies and counterstained with Phalloidin-TRITC. Bar, 100 μm. Representative images of three independent experiments.

Supplementary Figure 3 Regulation of Axl and Mer by steroid hormones.

BMDM cultures were stimulated with 1 μM Dex, hydrocortisone, cortisone, aldosterone, 17β-estradiol, estrone, estriol or progesterone for 24 h. Axl and Mer expression was assayed by immunoblotting. Representative of two independent experiments.

Supplementary Figure 4 Anti-inflammatory effects of Dex are independent of Mer and Axl.

(a) RT-PCR showing the kinetics of Mertk, Fpr1, Mrc1 mRNA induction and Axl and Il21r mRNA inhibition in BMDMs in response to 0.1 μM Dex. (b) BMDMs from indicated mice were treated with 100 ng/ml LPS with or without 0.1 μM Dex. TNF secretion was measured 24 h later by ELISA in culture supernatants. Representative of three independent experiments. (c) RT-PCR showing changes in Tnf, Mfge8, Il21r and Mrc1 mRNAs in BMDMs after 24 h incubation with 0.1 μM Dex. Data in (a) and (c) are normalized to Hprt mRNA and presented as fold change relative to untreated cells. Average of two independent experiments, each done in technical duplicate, graphed as mean ± s.d. (d) Immunoblot showing changes in activity of Akt, ERK1/2 and p38 signaling pathways in response to 0.1 μM Dex treatment of BMDMs derived from indicated knock-out mice. Representative of two independent experiments.

Supplementary Figure 5 Induction of Axl by poly(I:C) and IFN-α in BMDMs.

Cells were incubated with either poly(I:C) (1 μg/ml) or IFN-α (250 U/ml) for the indicated times in hours (h), and then blotted for total Axl (top), Mer (middle), or GAPDH (bottom). Representative of two independent experiments.

Supplementary Figure 6 Flow cytometry–based phagocytosis assay.

Apoptotic cells are labeled with pH-sensitive dye, pHrodo. Once engulfed into the acidic environment of phagosomes, pHrodo fluorescence is enhanced and phagocytic macrophages are distinguished based on their side scatter (SSC-A) and pHrodo fluorescence intensity using flow cytometry. In this experiment, the percent cells in the phagocytic gate is quantified in an 1-hour assay, in absence or presence of 10 nM GAS-6. Representative plot of 6 independent experiments.

Supplementary Figure 7 Regulation of the expression of Axl and Mer and phagocytosis in BMDCs.

(a) BMDC cultures from the indicated mice were stimulated for 10 min with 10 nM GAS-6 (G) or 25 nM Protein S (S). Receptor activation was assayed by immunoprecipitation and immunoblotting. Representative of two independent experiments. (b) BMDCs were cultured for 18 h in the presence of 0.1 μM Dex or 10 μg/ml poly(I:C) and then stimulated for 10 min with 10 nM GAS-6. Receptor activation was assayed by immunoprecipitation and immunoblotting. Representative of two independent experiments. (c, d) BMDCs from mice of the indicated genotypes were cultured for 24 h in the presence of 0.1 μM Dex (c) or 10 μg/ml poly(I:C) (d) and then incubated for 1 h with pHrodo stained ACs with or without 10 nM GAS-6. Percent of phagocytosis was measured using flow cytometry. Data are presented as mean ± s.d. from two independent experiments, each done for duplicate cultures for each condition.

Supplementary Figure 8 Model for the differences in the regulation and action of Axl and Mer in inflammatory and tolerogenic environments.

See text for details.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Table 1 (PDF 11409 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zagórska, A., Través, P., Lew, E. et al. Diversification of TAM receptor tyrosine kinase function. Nat Immunol 15, 920–928 (2014). https://doi.org/10.1038/ni.2986

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.2986

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing